Aga. Practice Update Barrett S Esophagus. 2022

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Clinical Gastroenterology and Hepatology 2022;20:2696–2706

CLINICAL PRACTICE UPDATES


AGA Clinical Practice Update on New Technology and
Innovation for Surveillance and Screening in Barrett’s
Esophagus: Expert Review
V. Raman Muthusamy,1 Sachin Wani,2 C. Prakash Gyawali,3 and
Srinadh Komanduri,4 for the CGIT Barrett’s Esophagus Consensus Conference
Participants
1
Vatche and Tamar Manoukian Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, California;
2
Division of Gastroenterology and Hepatology, University of Colorado School of Medicine, Denver, Colorado; 3Division of
Gastroenterology, Washington University School of Medicine, St. Louis, Missouri; and 4Division of Gastroenterology and
Hepatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois

DESCRIPTION: The purpose of this best practice advice (BPA) article from the Clinical Practice Update Com-
mittee of the American Gastroenterological Association is to provide an update on advances and
innovation regarding the screening and surveillance of Barrett’s esophagus.

METHODS: The BPA statements presented here were developed from expert review of existing literature
combined with discussion and expert opinion to provide practical advice. Formal rating of the
quality of evidence or strength of BPAs was not the intent of this clinical practice update. This
expert review was commissioned and approved by the AGA Institute Clinical Practice Updates
Committee (CPUC) and the AGA Governing Board to provide timely guidance on a topic of high
clinical importance to the AGA membership, and underwent internal peer review by the CPUC and
external peer review through standard procedures of Clinical Gastroenterology and Hepatology.

BEST PRACTICE Screening with standard upper endoscopy may be considered in individuals with at least 3
ADVICE 1: established risk factors for Barrett’s esophagus (BE) and esophageal adenocarcinoma,
including individuals who are male, non-Hispanic white, age >50 years, have a history of
smoking, chronic gastroesophageal reflux disease, obesity, or a family history of BE or esoph-
ageal adenocarcinoma.

BEST PRACTICE Nonendoscopic cell-collection devices may be considered as an option to screen for BE.
ADVICE 2:
BEST PRACTICE Screening and surveillance endoscopic examination should be performed using high-definition
ADVICE 3: white light endoscopy and virtual chromoendoscopy, with endoscopists spending adequate time
inspecting the Barrett’s segment.

BEST PRACTICE Screening and surveillance exams should define the extent of BE using a standardized
ADVICE 4: grading system documenting the circumferential and maximal extent of the columnar lined
esophagus (Prague classification) with a clear description of landmarks and the location and
characteristics of visible lesions (nodularity, ulceration), when present.

BEST PRACTICE Advanced imaging technologies such as endomicroscopy may be used as adjunctive techniques
ADVICE 5: to identify dysplasia.

BEST PRACTICE Sampling during screening and surveillance exams should be performed using the Seattle bi-
ADVICE 6: opsy protocol (4-quadrant biopsies every 1–2 cm and target biopsies from any visible lesion).

Abbreviations used in this paper: AGA, American Gastroenterological Kingdom; US, United States; VC, virtual chromoendoscopy; WATS-3D,
Association; BE, Barrett’s esophagus; BPA, Best Practice Advice; CGIT, wide-area transepithelial sampling.
Center for GI Innovation and Technology; CI, confidence interval; CLE,
confocal laser endomicroscopy; EAC, esophageal adenocarcinoma; EET, Most current article
endoscopic eradication therapy; GERD, gastroesophageal reflux disease;
HD-WLE, high definition-white light endoscopy; HGD, high-grade © 2022 by the AGA Institute
dysplasia; LGD, low-grade dysplasia; NDBE, nondysplastic Barrett’s 1542-3565/$36.00
esophagus; OR, odds ratio; PPI, proton pump inhibitor; UK, United https://doi.org/10.1016/j.cgh.2022.06.003

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
December 2022 Screening and Surveillance of Barrett’s Esophagus 2697

BEST PRACTICE Wide-area transepithelial sampling may be used as an adjunctive technique to sample the
ADVICE 7: suspected or established Barrett’s segment (in addition to the Seattle biopsy protocol).

BEST PRACTICE Patients with erosive esophagitis should be biopsied when concern of dysplasia or malignancy
ADVICE 8: exists. A repeat endoscopy should be performed after 8 weeks of twice a day proton pump
inhibitor therapy.

BEST PRACTICE Tissue systems pathology-based prediction assay may be utilized for risk stratification of pa-
ADVICE 9: tients with nondysplastic BE.

BEST PRACTICE Risk stratification models may be utilized to selectively identify individuals at risk for Barrett’s
ADVICE 10: associated neoplasia.

BEST PRACTICE Given the significant interobserver variability among pathologists, the diagnosis of BE-related
ADVICE 11: neoplasia should be confirmed by an expert pathology review.

BEST PRACTICE Patients with BE-related neoplasia should be referred to endoscopists with expertise in
ADVICE 12: advanced imaging, resection, and ablation.

BEST PRACTICE All patients with BE should be placed on at least daily proton pump inhibitor therapy.
ADVICE 13:
BEST PRACTICE Patients with nondysplastic BE should undergo surveillance endoscopy in 3 to 5 years.
ADVICE 14:
BEST PRACTICE In patients undergoing surveillance after endoscopic eradication therapy, random biopsies
ADVICE 15: should be taken of the esophagogastric junction, gastric cardia, and the distal 2 cm of the
neosquamous epithelium as well as from all visible lesions, independent of the length of the
original BE segment.

ndoscopic screening for Barrett’s esophagus (BE) discuss current needs and gaps in innovation relevant to
E and subsequent surveillance is supported by
current societal guidelines based on the potential for
the topic. This was a comprehensive didactic and dis-
cussion session created to provide a novel interactive
early detection of BE, dysplasia and neoplasia, and the environment to foster the AGA CGIT mission. The topic of
option for endoscopic eradication therapy (EET), with an this clinical practice update was thoroughly discussed by
overarching goal of reducing the morbidity and mortality expert faculty contributors selected by AGA CGIT, in-
of esophageal adenocarcinoma (EAC).1,2 However, less dustry representatives, and the patient advocate at the
than 20% of patients diagnosed with EAC in the United conference organized and hosted by AGA CGIT. The
States (US) have a preceding diagnosis of BE,3 suggesting content of this expert review was generated, discussed,
that current screening paradigms are inadequate. and voted upon by the expert faculty contributors at a
The purpose of this best practice advice article from closed-door meeting during the AGA CGIT conference. All
the Clinical Practice Update Committee of the American faculty contributors provided up to date declaration of
Gastroenterological Association (AGA) is to provide an conflicts of interest to ensure credibility of this docu-
update on advances and innovation regarding the ment, and signed off on the final manuscript, which un-
screening and surveillance of BE. The target audience is derwent internal peer review by the Clinical Practice
all gastroenterologists and endoscopists, and the target Updates Committee as well as external peer review
patient population is adults with known or suspected BE. through standard procedures of Clinical Gastroenterology
and Hepatology.
Methods
Screening for Barrett’s Esophagus
This expert review was commissioned jointly by the
AGA Institute Clinical Practice Updates Committee, the Best Practice Advice 1: Screening with standard upper
AGA Center for GI Innovation and Technology (CGIT), endoscopy may be considered in individuals with at least 3
and the AGA Governing Board to provide timely guidance established risk factors for BE and EAC, including in-
on a topic of high clinical importance to the AGA mem- dividuals who are male, non-Hispanic white, age >50
bership. The AGA CGIT Consensus Conferences bring years, have a history of smoking, chronic gastroesophageal
together content experts, stakeholders (industry, regu- reflux disease (GERD), obesity, or a family history of BE or
latory, and payors), along with a patient advocate to EAC.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
2698 Muthusamy et al Clinical Gastroenterology and Hepatology Vol. 20, No. 12

Current guidelines suggest endoscopic screening for population. Future studies should assess the impact of
an “at-risk” population.2 The vast majority of patients this approach (benefits and harms) in screening for BE.
(up to 90%) with EAC have never had a diagnosis of BE. Best Practice Advice 2: Nonendoscopic cell-collection
A recent meta-analysis of 49 studies suggests that the devices can be considered as an option to screen for BE.
prevalence of BE in the GERD population is 3%, and this Although upper endoscopy with biopsies remains the
increases with each additional risk factor.4 These risk gold standard for the diagnosis of BE, there is a signifi-
factors for BE include the presence of chronic GERD and cant need for noninvasive screening tools that are easy to
at least 2 of the following: age >50 years, male gender, administer, patient friendly, and cost-effective for the
Caucasian race, smoking, obesity, family history of BE or detection of BE. Transnasal endoscopy offers the use of
EAC.1 The highest prevalence was seen with family his- an ultrathin endoscope that can be performed in the
tory along with GERD at 23.4%.4 Chronic GERD is office setting without sedation. Although guidelines have
defined as >5 years or symptoms (heartburn or regur- acknowledged this as an alternative to sedated endos-
gitation) occurring frequently (weekly or greater). copy, it remains costly, expert-dependent, and not
However, the requirement of GERD symptoms has desirable to patients.2 This gap has led to the develop-
significantly limited the impact of screening on detection ment of multiple novel cell collection devices which offer
of EAC. a nonendoscopic alternative for screening. Current non-
The panel discussed the limitations of chronic GERD endoscopic cell collection devices include Cytosponge
symptom as a mandatory prerequisite for endoscopic (Medtronic GI Solutions), EsoCheck (Lucid Diagnostics),
screening. A recent study of prevalent EAC assessed so- and EsophaCap (Capnostics) (See Supplementary
cietal guidelines in the US (n ¼ 663) and United Kingdom Appendix). All 3 nonendoscopic devices have demon-
(UK) (n ¼ 645) to determine the sensitivity of current strated excellent tolerability, safety, and sensitivity for
screening recommendations.1,5,6 In these cohorts, 54.9% the diagnosis of BE. Further data is needed to validate
of the US patients and 38.9% of the UK patients would patient selection and the optimal setting for adminis-
not have been identified by current screening guidelines. tration of these novel devices in the US.
Furthermore, the reason most patients (US, 86.5%; UK,
61.4%) did not meet screening guidelines was the lack of Endoscopic Examination of Barrett’s
symptomatic GERD. Furthermore, a second study of US Esophagus
veterans also identified that >50% of patients with EAC
did not have frequent GERD symptoms and would not Best Practice Advice 3: Screening and surveillance
have met current screening guidelines.7 endoscopic examination should be performed using high-
Multiple predictive tools have been developed iden- definition white light endoscopy (HD-WLE) and virtual
tify patients at risk of BE. These tools not only use GERD chromoendoscopy (VC), with endoscopists spending
symptoms, but also multiple other clinical and de- adequate time inspecting the Barrett’s segment.
mographic factors implicated in BE and EAC.8 These The goal of endoscopic screening and surveillance in
predictive indices have been developed for real-time risk BE is early detection of BE-related dysplasia and early
assessment without a prerequisite of GERD through pa- EAC. Consistent with recent guidelines,2,9 the panel
tient assessment and questionnaires. From the available agrees with the routine use of HD-WLE and VC during
tools, the HUNT (Nord-Trondelag Health Study), M- screening and surveillance endoscopy in patients with
BERET (Michigan BE pREdiction Tool), and Kunzmann BE. In an updated meta-analysis that included 504 pa-
tools were found to be more sensitive for predicting BE tients, virtual chromoendoscopy with HD-WLE was
than GERD symptoms alone.8 Although further validation associated with a higher detection rate of HGD/EAC
of these tools is needed, they can be considered in the compared with HD-WLE alone (14.7% vs 10.1%; relative
clinical evaluation of patients for endoscopic screening. risk, 1.44).10 Although available data suggest comparable
The optimal number of risk factors for screening re- rates of dysplasia detection between virtual and tradi-
mains to be well-defined and inclusion of GERD remains tional chromoendoscopy techniques, VC is the preferred
fraught with several limitations. The threshold number approach as this imaging platform is available in most
of risk factors is largely based on expert opinion. These endoscopes, requires no additional costs, and circum-
factors provided the impetus to propose a screening vents the problems associated with dye-based chro-
approach that is not restricted to patients with GERD and moendoscopy such as the need for dye spraying
considers all defined risk factors for BE and EAC. equipment, additional time required, cumbersome na-
Therefore, screening with standard upper endoscopy ture of the procedure, difficulty in achieving uniform
may be considered in individuals with at least 3 estab- coating of the mucosal surface with the dye, and inability
lished risk factors for BE and EAC, including individuals to detect superficial vascular patterns.2 Incorporation of
who are male, non-Hispanic white, age >50 years, have a training in virtual and traditional chromoendoscopy
history of smoking, chronic GERD, obesity, or a family during fellowship and training programs for the prac-
history of BE or EAC. The reduction of the valuation of ticing endoscopists will be important for widespread
GERD in this paradigm is felt to significantly improve routine implementation in clinical practice.10 The clinical
detection of BE and more accurately identify the “at-risk” use of VC is suggested regardless of endoscope

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
December 2022 Screening and Surveillance of Barrett’s Esophagus 2699

Table 1. Ten-step Approach to Endoscopic Examination of Barrett’s Esophagus (BE)

Approach Rationale

1. Identify esophageal landmarks, including the location of the 1. Critical for future exams
diaphragmatic hiatus, gastroesophageal junction, and squa-
mocolumnar junction

2. Consider use of a distal attachment cap (especially in patients 2. Facilitate visualization


with prior diagnosis of dysplasia)

3. Clean mucosa well using water jet channel and carefully suction 3. Remove any distracting mucus or debris and
the fluid minimize mucosal trauma

4. Utilize insufflation and desufflation 4. Fine adjustments to luminal insufflation can


help with detection of subtle abnormalities

5. Spend adequate time inspecting the Barrett’s segment and 5. Careful examination increases dysplasia
gastric cardia in retroflexion detection

6. Examine the Barrett’s segment using HD-WLE 6. Standard of care

7. Examine the Barrett’s segment using chromoendoscopy 7. Enhances mucosa pattern and surface
(including virtual chromoendoscopy) vasculature

8. Use the Prague classification to describe the circumferential and 8. Standardized reporting system
maximal Barrett’s segment length

9. Use the Paris classification to describe superficial neoplasia 9. Standardized reporting system

10. Use the Seattle protocol (in conjunction with virtual chro- 10. Increases dysplasia detection
moendoscopy) with a partially deflated esophagus to sample
the Barrett’s segment

Adapted from Kolb J, Wani S. Curr Opin Gastroenterol 2020;36:351–358.60


HD-WLE, High-definition white light endoscopy.

manufacturer, but is should be clear that majority of data proportion of HGD or EAC are missed within the first
supporting this is for narrow-band imaging only. There year following the index endoscopy that diagnosed
are limited data addressing the impact of inspection time BE.18,19 To address the importance of the quality of
on detection of BE-associated neoplasia in patients un- endoscopic examination, using an evidence-based
dergoing screening and surveillance endoscopy.11-13 approach, an international working group recently
Conceptually, there was agreement that adequate in- standardized terminology and definitions for post-
spection time would lead to more careful examination of endoscopy esophageal adenocarcinoma and post-
the BE mucosa and potentially increased detection of BE- endoscopy esophageal neoplasia; EAC and HGD/EAC
associated neoplasia. Future studies need to define the detected before the next recommended surveillance
optimal threshold for inspection time per cm of the BE endoscopy in a patient with nondysplastic BE (NDBE),
segment. Although the panel purposefully did not name a respectively.10 A conceptual 10-step approach to a high-
time period comprising an adequate exam due to lack of quality endoscopic examination in patients with BE is
data on this issue, the European Society for Gastroin- highlighted in Table 1.
testinal Endoscopy and United European Gastroenter- Best Practice Advice 4: Screening and surveillance
ology recommend a procedure time of 7 minutes for endoscopic examinations should define the extent of BE
upper endoscopy and inspection time of 1 minute/cm using a standardized grading system documenting the
of the circumferential extent of the Barrett’s mucosa.14 circumferential and maximal extent of the columnar lined
Although screening and surveillance upper endos- esophagus (Prague Classification) with a clear description
copy may be effective in detecting dysplasia and curable of landmarks and location and characteristics of visible
EAC, it is imperfect. Similar to post-colonoscopy colo- lesions (nodularity, ulceration), when present.
rectal cancer,15 BE-associated high-grade dysplasia The panel acknowledged that the impact of use of
(HGD) and EAC can be diagnosed before the next rec- standardized grading criteria for BE length (Prague
ommended endoscopic evaluation after an upper classification) and visible lesions (Paris classification)
endoscopy that was negative for HGD or EAC.16,17 Meta- (Figure 1, A) on critical outcomes such as improved
analyses and cohort studies suggest that a high detection of BE-associated neoplasia has not been

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
2700 Muthusamy et al Clinical Gastroenterology and Hepatology Vol. 20, No. 12

Figure 1. A, The Prague classification for BE. B, Illustration of the Seattle biopsy protocol for performing surveillance in pa-
tients with NDBE.59 C, Illustration of a simplified protocol for performing random surveillance biopsies in patients status post
EET. Of note, all visible lesions in the cardia and tubular esophagus should be biopsied separately.57 EGJ, Esophagogastric
junction.

assessed. The panel suggested the routine use of these artificial intelligence.22,23 The panelists felt strongly this
classification practices as surrogates for performance of was an important area where further innovation is
a high-quality endoscopy exam. needed, but that the use of these techniques was not
Best Practice Advice 5: Advanced imaging technologies required for a high-quality exam and the data to date
may be used as adjunctive imaging techniques to identify did not support its routine use. However, the panel felt
dysplasia. these technologies were promising and carried potential
The panel were supportive of the need to have benefits in select cases and currently might be best
improved imaging technologies to better identify areas utilized in expert centers.
of dysplasia and early cancer. Technologies considered Best Practice Advice 6: Sampling during screening and
for this discussion included confocal (CLE) or volu- surveillance endoscopic examinations should be performed
metric laser endomicroscopy. A meta-analysis of 14 using the Seattle biopsy protocol (4-quadrant biopsies
studies of 789 patients with 4047 lesions found CLE every 1‒2 cm and target biopsies from any visible lesion).
had a per-lesion analysis pooled sensitivity and speci- The support for this structured biopsy protocol is
ficity of 77% (95% confidence interval [CI], 0.73–0.81) based on observational data suggesting that the use of
and 89% (95% CI, 0.87–0.90), respectively.20 A separate the Seattle biopsy protocol (Figure 1, B) is associated
meta-analysis of 5 studies involving 251 patients with a higher dysplasia detection rate (relative risk,
assessing within-patient comparisons of narrow band 2.75).10 The panel acknowledged that endoscopists can
imaging and CLE found the pooled additional detection meet this criterion if they prefer not to sample a visible
rate of CLE for per-lesion detection of neoplasia in pa- lesion and refer the patient for endoscopic resection.
tients with BE was 19.3% (95% CI, 0.05–0.33), but a Unfortunately, several studies have consistently demon-
comparable per-patient pooled sensitivity and speci- strated suboptimal adherence rates to the Seattle biopsy
ficity.21 Volumetric laser endomicroscopy, though not protocol.24-26 The odds of detecting dysplasia signifi-
currently available commercially, has introduced several cantly decreased with nonadherence to the Seattle bi-
new advances with regards to imaging in BE, including opsy protocol (odds ratio [OR], 0.53; 95% CI,
laser marking and the interpretation of imaging using 0.35–0.82).24 A recent analysis using a national quality

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
December 2022 Screening and Surveillance of Barrett’s Esophagus 2701

benchmarking registry that included 58,709 endoscopies follow-up esophagogastroduodenoscopy may reveal un-
showed that nearly 20% of endoscopies were not derlying Barrett’s in up to 10% to 12% of patients.34-36
adherent to the Seattle biopsy protocol, and that endo-
scopists were less adherent with increasing BE length;
with the odds of nonadherence increasing by 31% with Risk Stratification of Barrett’s
every 1-cm increase in length.26 Esophagus
Best Practice Advice 7: Wide-area transepithelial
sampling (WATS-3D) may be used as an adjunctive tech- Best Practice Advice 9: Tissue systems pathology-based
nique to sample the suspected or established Barrett’s prediction assay may be utilized for risk stratification of
segment (in addition to the Seattle biopsy protocol). patients with NDBE.
WATS-3D is a novel method that uses an abrasive Risk stratification among patients with NDBE has been
brush to sample larger surface areas of the esophagus. limited to clinical scoring systems. Recently, a tissue sys-
These specimens allow for analysis of large sheets of tems pathology assay (Tissue Cypher), commercially
cells while maintaining the 3-dimensional aspects of the available in the US, has been validated and demonstrated
tissue. The tissue is analyzed by software that uses to accurately risk stratify patients with NDBE (low, in-
convoluted neural networks to identify abnormal cells, termediate, high) for progression to HGD/EAC with a 4.7-
which are confirmed by an expert pathologist. A recent fold increased risk in patients stratified as high risk.37 The
systematic review and meta-analysis of 7 studies assay is performed on routine biopsies from the Barrett’s
demonstrated an incremental yield for dysplasia detec- segment and quantifies 9 protein-based biomarkers (p16,
tion of 7.2%.27 In addition, pathologic interpretation of p53, AMACR, HER2, Cytokeratin 20, CD68, COX-2, HIF-
these specimens has been shown to have less interob- 1alpha, and CD45Ro), along with nuclear morphology and
server variability with kappa of 0.86.28 As such, the tissue architecture. It is performed on formalin-fixed,
recent ASGE guidelines supported the use of WATS-3D in paraffin-embedded biopsies. The result is a numeric
addition to Seattle protocol in select patients (eg, inde- score from 1 to 10 that corresponds to a patient’s risk for
terminate for dysplasia or clinically high-risk NDBE) progression. To date, there have been 5 independent
undergoing surveillance.2 Further prospective studies studies including 239 progressors and 656 non-
directly comparing WATS-3D and Seattle protocol are progressors across the US and Europe. The sensitivity and
needed to understand if WATS-3D sampling might be as specificity for Tissue cypher for detecting progression in
or more effective. patients with NDBE is 30.4% and 95%, respectively, with
Best Practice Advice 8: Patients with erosive esopha- the sensitivity increasing to 50% if multiple levels are
gitis may be biopsied when concern of dysplasia or ma- examined.38 A recent spatial-temporal analysis of pro-
lignancy exists, with the caveat that a repeat endoscopy gressors and nonprogressors demonstrated that a high-
after 8 weeks of twice-daily proton pump inhibitor (PPI) is risk score was associated with a rate of progression of
performed. 6.9%, similar to LGD.38 In addition, a study using Markov
The panel discussed the importance of preventing modeling suggesting that Tissue Cypher-based risk strat-
delays in diagnosing dysplasia and malignancy when ification becomes cost-effective after 5 years, with an in-
concerning endoscopic findings are encountered in the cremental cost-effectiveness ratio of $52,483/quality-
setting of esophagitis. Although the potential for over- adjusted life years. Finally, a recent pooled analysis of
calling dysplasia (especially low-grade dysplasia [LGD]) in international studies in 472 patients with NDBE demon-
the setting of active inflammation exists, the panel felt that strated that a high Tissue Cypher risk score was a strong
this should not preclude obtaining biopsies as expert pa- independent predictor for progression to HGD/EAC (OR,
thologists have been shown to be able to distinguish 14.2; 95% CI, 5–39; P < .001).39 Based on these data, the
inflammation from true LGD.29 When such samples are panel agreed that the Tissue Cypher assay may be of
obtained, documentation should be included regarding benefit for risk stratification of patients with NDBE.
the presence and severity of the esophagitis visualized. Best Practice Advice 10: Risk stratification models may
Although once a day PPI may be sufficient for healing be utilized to selectively identify individuals at risk for
some patients, given the potential for an incomplete Barrett’s associated neoplasia.
response in a subset of patients with severe esophagitis, The panel agreed on the value of using risk stratifi-
twice a day therapy was suggested to maximize efficacy, cation models to stratify surveillance intervals and in-
especially given the limited downside to such a short fluence the decision on whether to perform EET.
treatment course. Treatment for 8 weeks was suggested, However, at present, the only validated clinical risk
as this has been the typical duration of most trials of PPI stratification model for predicting progression to HGD/
for the healing of esophagitis and has been recommended esophageal cancer in patients with known BE is the
by prior guidelines.30-32 The panel noted that a relook Progression in Barrett’s Esophagus score.40 This score
endoscopy is only needed for those with Los Angeles was developed from 2697 patients, of whom 154 (5.7%)
Grade C and D esophagitis.33 The indication for repeat developed HGD or esophageal cancer. Factors signifi-
endoscopy is to document healing of esophagitis and to cantly associated with progression included baseline
assess for any features of malignancy. Furthermore, confirmed LGD, male sex, smoking, and BE length. Scores

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
2702 Muthusamy et al Clinical Gastroenterology and Hepatology Vol. 20, No. 12

Figure 2. Suggested BE care pathway.

assigned identified patients with BE that progressed to among pathologists and the importance of high-quality
HGD or EAC with a c-statistic of 0.76 (95% CI, 0.72–0.80; expert pathology review in the diagnosis of BE-related
P < .001), with the high-risk group progressing at a rate neoplasia.2,47 An accurate diagnosis of dysplasia is crit-
of 2.1% compared with 0.73% for the intermediate ical for clinical decision-making and risk stratification,
group and 0.13% for the low-risk group. Of note, this including the selection of endoscopic eradication therapy
score is heavily influenced by the presence of LGD but vs intensive surveillance. A systematic review and meta-
was found to perform better in predicting progression analysis showed that expert pathology review resulted in
than LGD alone in a separate BE cohort.41 Other non- a change in the pathologic diagnosis (upgrading or
validated models using clinical variables have included downgrading) in 55% (95% CI, 31%–77%) of all pa-
the presence of esophagitis, lack of PPI use, being over- tients.47 Available data suggests LGD, as confirmed by
weight, increasing age, and a known duration of BE of expert pathology review, is associated with a higher rate
10 years.42-46 The panelists noted that several addi- of disease progression to HGD/EAC.47 P53 immunohis-
tional models were currently in development and noted tochemistry can help confirm dysplasia and improve
that models incorporating both clinical and biomarker consistency of reporting.48,49 This Best Practice Advice is
parameters would likely ultimately be needed to opti- consistent with GI guidelines that recommend confir-
mize predictive accuracy. mation of dysplasia of any grade by a second pathologist
with expertise in GI pathology.9,47
Provider Expertise in Managing BE Best Practice Advice 12: Patients with BE-related
neoplasia should be referred to endoscopists with exper-
Best Practice Advice 11: Given the significant interob- tise in advanced imaging, resection, and ablation.
server variability among pathologists, the diagnosis of Physicians with expertise in Barrett’s neoplasia man-
Barrett’s-related neoplasia should be confirmed by an agement have been shown to identify more visible lesions
expert pathology review. compared with nonexperts.50 The panelists strongly felt
The panelists acknowledged the significant interob- that physicians performing endoscopic eradication therapy
server variability in the interpretation of dysplasia for BE-related neoplasia should either perform or work in

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
December 2022 Screening and Surveillance of Barrett’s Esophagus 2703

centers that can offer both resection and ablation tech- risk of several outcomes.54,55 Evidence is inadequate to
niques, as recommended by prior quality metrics.51 Expert establish causal relationships between PPI and any of
centers should ideally be defined based on adequate vol- these proposed associations, with the exception of
ume, availability of needed technology, procedural exper- enteric infection.55 Given the unclear benefit of higher
tise, and exceeding established quality metrics. doses of PPI on oncogenesis, the panel suggested at least
daily dosing, with higher doses considered for those
requiring them for symptom control and among patients
Follow-up and Surveillance of BE with BE-related neoplasia undergoing endoscopic eradi-
cation therapy.
Best Practice Advice 13: Patients with BE should be Best Practice Advice 14: Patients with NDBE should
placed on at least daily PPI therapy. undergo surveillance endoscopy in 3 to 5 years.
Epidemiologic evidence from observational studies Endoscopic surveillance in patients with known BE
that have demonstrated a significant decrease in the risk remains the gold standard for dysplasia and neoplasia
of progression to HGD and EAC in patients with BE with detection. Current guidelines recommend endoscopic
PPI therapy. A systematic review and meta-analysis surveillance every 3 to 5 years1 based on a low risk of
showed that PPI therapy was associated with a 71% progression to HGD/EAC in patients with NDBE. Surveil-
reduction in the risk of HGD or EAC (adjusted OR, 0.29; lance intervals are shortened significantly in patients with
95% CI, 0.12–0.79).52 In 4 cohort studies that reported dysplasia but should remain 3 to 5 years for patients with
the time to progression to HGD or EAC, PPI users were NDBE. The interval allows for gastroenterologists some
also significantly less likely to progress to HGD or EAC flexibility to individualize intervals for each patient. Most
(adjusted hazard ratio, 0.32; 95% CI, 0.15–0.67). The recently, the American College of Gastroenterology
AspECT trial demonstrated that high-dose PPI was su- guidelines for 2022 recommended consideration of the
perior to low-dose PPI for lengthening the time to reach segment length when determining surveillance interval,
the combined end point of death from any cause, EAC, or with longer intervals for segments <3 cm.9 Careful dis-
HGD. However, several study limitations prevent these cussions and assessments of the value of endoscopic sur-
conclusions to be generalizable. The trial was not double- veillance, given other comorbidity and risks, should be a
blinded, the event rate was low, and only a small effect part of the management of all patients with BE.
size was noted. The overall benefit was skewed towards Best Practice Advice 15: In patients undergoing sur-
all cause-mortality rather than cancer-related mortality, veillance after endoscopic eradication therapy (EET),
most relevant to the BE population.53 As such, there was 4-quadrant random biopsies should be taken of the esoph-
insufficient information in these studies on whether agogastric junction, gastric cardia, and the distal 2 cm of
taking PPI twice daily would provide any added benefit the neosquamous epithelium, as well as from all visible le-
over once daily administration. The panel also consid- sions, independent of the length of the original BE segment.
ered the potential harms of long-term PPI therapy and Traditionally, 4-quadrant random post-EET sur-
the suggested associations between PPI therapy and the veillance biopsies have been recommended in the

Best Practice Advice (BPA) Statements

Screening for Barrett’s Esophagus (BE)


BPA #1. Screening with standard upper endoscopy may be considered in individuals with established risk factors for BE and esophageal
adenocarcinoma – presence of at least 3 risk factors (individuals who are male, non-Hispanic white, age >50 years, have a history of
smoking, chronic gastrointestinal reflux disease, obesity, or a family history of BE or esophageal adenocarcinoma).
BPA #2. Nonendoscopic cell collection devices can be considered as an option to screen for BE.
Endoscopic Examination of BE
BPA #3. Screening and surveillance exams should be performed using high-definition white light endoscopy and virtual chromoendoscopy,
with endoscopists spending adequate time inspecting the Barrett’s segment.
BPA #4. Screening and surveillance exams should define the extent of BE using a standardized grading system documenting the
circumferential and maximal extent of the columnar lined esophagus (Prague classification) with a clear description of landmarks and the
location and characteristics of visible lesions (nodularity, ulceration), when present.
BPA #5. Advanced imaging technologies such as endomicroscopy may be used as adjunctive imaging techniques to identify dysplasia.
BPA #6. Sampling during screening and surveillance exams should be performed using the Seattle biopsy protocol (4-quadrant biopsies
every 1‒2 cm and target biopsies from any visible lesion).
BPA #7. Wide area transepithelial sampling may be used as an adjunctive technique to sample the suspected or established Barrett’s
segment (in addition to the Seattle biopsy protocol).
BPA #8. Patients with erosive esophagitis may be biopsied when concern of dysplasia or malignancy exists, with the caveat that a repeat
endoscopy after 8 weeks of twice a day proton pump inhibitors is performed.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
2704 Muthusamy et al Clinical Gastroenterology and Hepatology Vol. 20, No. 12

Continued

Best Practice Advice (BPA) Statements


Risk Stratification of BE
BPA #9. Tissue systems pathology-based prediction assay may be utilized for risk stratification of patients with nondysplastic BE.
BPA #10. Risk stratification models may be utilized to selectively identify individuals at risk for Barrett’s associated neoplasia.
Provider Expertise in Managing BE
BPA #11. Given the significant interobserver variability among pathologists, the diagnosis of BE-related neoplasia should be confirmed by an
expert pathology review.
BPA #12. Patients with BE-related neoplasia should be referred to endoscopists with expertise in advanced imaging, resection, and ablation.
Follow-up and Surveillance of BE
BPA #13. Patients with BE should be placed on at least daily proton pump inhibitor therapy.
BPA #14. Patients with nondysplastic BE should undergo surveillance endoscopy in 3 to 5 years.
BPA #15. In patients undergoing surveillance after endoscopic eradication therapy, 4-quadrant random biopsies should be taken of the
esophagogastric junction, gastric cardia, and the distal 2 cm of the neosquamous epithelium as well as from all visible lesions,
independent of the length of the original BE segment.

cardia, at the esophagogastric junction and every 1 References


cm in entire prior BE segment. Three studies have 1. Shaheen NJ, Falk GW, Iyer PG, et al. American College of
reported on the anatomic location of recurrent BE Gastroenterology. ACG Clinical Guideline: diagnosis and man-
after EET.56-58 In total, they evaluated 1235 patients agement of Barrett’s esophagus. Am J Gastroenterol 2016;
achieving complete eradication of intestinal meta- 111:30–50; quiz: 51.
plasia and observed 233 recurrences, for an aggregate 2. ASGE Standards of Practice Committee, Qumseya B, Sultan S,
recurrence rate of 18.9%. The majority of nonvisible Bain P, et al. ASGE guideline on screening and surveillance of
recurrence of intestinal metaplasia occurred at the Barrett’s esophagus. Gastrointest Endosc 2019;90:335–359.e2.
esophagogastric junction, whereas most recurrences 3. Tan MC, Mansour N, White DL, et al. Systematic review with
meta-analysis: prevalence of prior and concurrent Barrett’s
in the tubular esophagus were visible. Using the
oesophagus in oesophageal adenocarcinoma patients. Aliment
aforementioned best practice statement post-EET
Pharmacol Ther 2020;52:20–36.
surveillance biopsy strategy, 98% (228/233) of all
4. Qumseya BJ, Bukannan A, Gendy S, et al. Systematic review
recurrences could be identified. The panelists felt that and meta-analysis of prevalence and risk factors for Barrett’s
for a BE length of <2 cm, such an approach was still esophagus. Gastrointest Endosc 2019;90:707–717.e1.
reasonable given the potential for subsquamous/ 5. Sawas T, Zamani SA, Killcoyne S, et al. Limitations of heartburn
tangential extension up to 1 cm beyond the proximal and other societies’ criteria in Barrett’s screening for detecting
end of the squamocolumnar junction. The panelists de novo esophageal adenocarcinoma. Clin Gastroenterol Hep-
also recognized the value of obtaining cardia biopsies atol 2021 (Online ahead of print).
to assess for dysplasia (Figure 1, C). 6. Fitzgerald RC, di Pietro M, Ragunath K, et al. , British Society of
Gastroenterology. British Society of Gastroenterology guide-
lines on the diagnosis and management of Barrett’s oesoph-
Conclusion agus. Gut 2014;63:7–42.
7. Nguyen TH, Thrift AP, Rugge M, et al. Prevalence of Barrett’s
Targeted BE screening and surveillance using existing esophagus and performance of societal screening guidelines in
methodologies, as well as use of emerging and novel an unreferred primary care population of U.S. veterans. Gas-
screening technologies, have the potential to improve trointest Endosc 2021;93:409–419.e1.
early detection of dysplasia, neoplasia, and EAC within 8. Rubenstein JH, McConnell D, Waljee AK, et al. Validation and
populations at risk for BE (Figure 2). comparison of tools for selecting individuals to screen for Bar-
rett’s esophagus and early neoplasia. Gastroenterology 2020;
158:2082–2092.
Supplementary Material 9. Shaheen NJ, Falk GW, Iyer P, et al. Diagnosis and management
of Barrett’s esophagus: an updated ACG Guideline. Am J
Note: To access the supplementary material accom- Gastroenterol 2022;117:559–587.
panying this article, visit the online version of Clinical 10. Wani S, Yadlapati R, Singh S, et al. Post-Endoscopy Esopha-
Gastroenterology and Hepatology at www.cghjournal.org, geal Neoplasia Expert Consensus Panel. Postendoscopy
and at https://doi.org/10.1016/j.cgh.2022.06.003. esophageal neoplasia in Barrett’s esophagus: consensus

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
December 2022 Screening and Surveillance of Barrett’s Esophagus 2705

statements from an international expert panel. Gastroenterology esophagus: an analysis of biopsy sampling practices from a
2021;162:366–372. quality improvement registry. Gastrointest Endosc 2019;
11. Gupta N, Gaddam S, Wani SB, et al. Longer inspection time is 90:732–741.e3.
associated with increased detection of high-grade dysplasia 27. Codipilly DC, Krishna Chandar A, Wang KK, et al. Wide-area
and esophageal adenocarcinoma in Barrett’s esophagus. Gas- transepithelial sampling for dysplasia detection in Barrett’s
trointest Endosc 2012;76:531–538. esophagus: a systematic review and meta-analysis. Gastro-
12. Kawamura T, Wada H, Sakiyama N, et al. Examination time as a intest Endosc 2022;95:51–59.e7.
quality indicator of screening upper gastrointestinal endoscopy 28. Vennalaganti P, Kanakadandi V, Goldblum JR, et al. Discor-
for asymptomatic examinees. Dig Endosc 2017;29:569–575. dance among pathologists in the United States and Europe in
13. Park JM, Huo SM, Lee HH, et al. Longer observation time in- diagnosis of low-grade dysplasia for patients with Barrett’s
creases proportion of neoplasms detected by esophagogas- esophagus. Gastroenterology 2017;152:564–570.e4.
troduodenoscopy. Gastroenterology 2017;153:460–469.e1. 29. Curvers WL, ten Kate FJ, Krishnadath KK, et al. Low-grade
14. Bisschops R, Areia M, Coron E, et al. Performance measures for dysplasia in Barrett’s esophagus: overdiagnosed and under-
upper gastrointestinal endoscopy: a European Society of estimated. Am J Gastroenterol 2010;105:1523–1530.
Gastrointestinal Endoscopy (ESGE) Quality Improvement Initia- 30. Castell DO, Kahrilas PJ, Richter JE, et al. Esomeprazole (40 mg)
tive. Endoscopy 2016;48:843–864. compared with lansoprazole (30 mg) in the treatment of erosive
15. Rutter MD, Beintaris I, Valori R, et al. World Endoscopy Orga- esophagitis. Am J Gastroenterol 2002;97:575–583.
nization consensus statements on post-colonoscopy and post- 31. ASGE Standards of Practice Committee, Muthusamy VR,
imaging colorectal cancer. Gastroenterology 2018; Lightdale JR, Acosta RD, et al. The role of endoscopy in the man-
155:909–925.e3. agement of GERD. Gastrointest Endosc 2015;81:1305–1310.
16. Duloy A, Keswani R, Hall M, et al. Time given to trainees to attempt 32. Richter JE, Kahrilas PJ, Sontag SJ, et al. Comparing lansopra-
cannulation during endoscopic retrograde cholangiopancreatog- zole and omeprazole in onset of heartburn relief: results of a
raphy varies by training program and is not associated with randomized, controlled trial in erosive esophagitis patients. Am
competence. Clin Gastroenterol Hepatol 2020;18:3040–3042.e1. J Gastroenterol 2001;96:3089–3098.
17. Holmberg D, Ness-Jensen E, Mattsson F, et al. Adherence to 33. Katz PO, Dunbar KB, Schnoll-Sussman FH, et al. ACG
clinical guidelines for Barrett’s esophagus. Scand J Gastro- Clinical Guideline for the diagnosis and management of
enterol 2019;54:945–952. gastroesophageal reflux disease. Am J Gastroenterol 2022;
18. Sawas T, Majzoub AM, Haddad J, et al. Magnitude and time- 117:27–56.
trend analysis of post-endoscopy esophageal adenocarci- 34. Hanna S, Rastogi A, Weston AP, et al. Detection of Barrett’s
noma: a systematic review and meta-analysis. Clin Gastro- esophagus after endoscopic healing of erosive esophagitis. Am
enterol Hepatol 2022;20:e31–e50. J Gastroenterol 2006;101:1416–1420.
19. Vajravelu RK, Kolb JM, Thanawala SU, et al. Characterization of 35. Modiano N, Gerson LB. Risk factors for the detection of Bar-
prevalent, post-endoscopy, and incident esophageal cancer in rett’s esophagus in patients with erosive esophagitis. Gastro-
the United States: a large retrospective cohort study. Clin intest Endosc 2009;69:1014–1020.
Gastroenterol Hepatol 2021 (Online ahead of print). 36. Rodriguez S, Mattek N, Lieberman D, et al. Barrett’s esophagus
20. Xiong YQ, Ma SJ, Zhou JH, et al. A meta-analysis of confocal on repeat endoscopy: should we look more than once? Am J
laser endomicroscopy for the detection of neoplasia in patients Gastroenterol 2008;103:1892–1897.
with Barrett’s esophagus. J Gastroenterol Hepatol 2016; 37. Davison JM, Goldblum J, Grewal US, et al. Independent blinded
31:1102–1110. validation of a tissue systems pathology test to predict pro-
21. Xiong YQ, Ma SJ, Hu HY, et al. Comparison of narrow-band gression in patients with Barrett’s esophagus. Am J Gastro-
imaging and confocal laser endomicroscopy for the detection enterol 2020;115:843–852.
of neoplasia in Barrett’s esophagus: a meta-analysis. Clin Res 38. Frei NF, Konte K, Bossart EA, et al. Independent validation of a
Hepatol Gastroenterol 2018;42:31–39. tissue systems pathology assay to predict future progression in
22. Swager AF, de Groof AJ, Meijer SL, et al. Feasibility of laser nondysplastic Barrett’s esophagus: a spatial-temporal analysis.
marking in Barrett’s esophagus with volumetric laser endomi- Clin Transl Gastroenterol 2020;11:e00244.
croscopy: first-in-man pilot study. Gastrointest Endosc 2017; 39. Iyer P, Codipilly DC, Chandar A, et al. Prediction of progression
86:464–472. in Barrett’s esophagus using a tissue systems pathology test: a
23. Struyvenberg MR, de Groof AJ, Fonolla R, et al. Prospective pooled analysis of international studies. Clin Gastroenterol
development and validation of a volumetric laser endomicro- Hepatol 2022 (Online ahead of print).
scopy computer algorithm for detection of Barrett’s neoplasia. 40. Parasa S, Vennalaganti S, Gaddam S, et al. Development and
Gastrointest Endosc 2021;93:871–879. validation of a model to determine risk of progression of Bar-
24. Abrams JA, Kapel RC, Lindberg GM, et al. Adherence to biopsy rett’s esophagus to neoplasia. Gastroenterology 2018;
guidelines for Barrett’s esophagus surveillance in the commu- 154:1282–1289.e2.
nity setting in the United States. Clin Gastroenterol Hepatol 41. Kunzmann AT, Thrift AP, Johnston BT, et al. External validation
2009;7:736–742; quiz: 710. of a model to determine risk of progression of Barrett’s
25. Westerveld D, Khullar V, Mramba L, et al. Adherence to quality oesophagus to neoplasia. Aliment Pharmacol Ther 2019;
indicators and surveillance guidelines in the management of 49:1274–1281.
Barrett’s esophagus: a retrospective analysis. Endosc Int Open 42. Sikkema M, Looman CW, Steyerberg EW, et al. Predictors for
2018;6:E300–E307. neoplastic progression in patients with Barrett’s esophagus: a
26. Wani S, Williams JL, Komanduri S, et al. Endoscopists sys- prospective cohort study. Am J Gastroenterol 2011;
tematically undersample patients with long-segment Barrett’s 106:1231–1238.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
2706 Muthusamy et al Clinical Gastroenterology and Hepatology Vol. 20, No. 12

43. Krishnamoorthi R, Borah B, Heien H, et al. Rates and predictors 54. Vaezi MF, Yang YX, Howden CW. Complications of proton
of progression to esophageal carcinoma in a large population- pump inhibitor therapy. Gastroenterology 2017;153:35–48.
based Barrett’s esophagus cohort. Gastrointest Endosc 2016; 55. Moayyedi P, Eikelboom JW, Bosch J, et al. COMPASS In-
84:40–46.e7. vestigators. Safety of proton pump inhibitors based on a large,
44. Holmberg D, Ness-Jensen E, Mattsson F, et al. Clinical pre- multi-year, randomized trial of patients receiving rivaroxaban or
diction model for tumor progression in Barrett’s esophagus. aspirin. Gastroenterology 2019;157:682–691.e2.
Surg Endosc 2019;33:2901–2908. 56. Cotton CC, Wolf WA, Pasricha S, et al. Recurrent intestinal
45. Brown CS, Lapin B, Goldstein JL, et al. Predicting progression in metaplasia after radiofrequency ablation for Barrett’s esoph-
Barrett’s esophagus: development and validation of the Bar- agus: endoscopic findings and anatomic location. Gastrointest
rett’s Esophagus Assessment of Risk Score (BEAR Score). Ann Endosc 2015;81:1362–1369.
Surg 2018;267:716–720. 57. Omar M, Thaker AM, Wani S, et al. Anatomic location of Bar-
46. Hoefnagel SJM, Mostafavi N, Timmer MR, et al. A genomic rett’s esophagus recurrence after endoscopic eradication ther-
biomarker-based model for cancer risk stratification of non- apy: development of a simplified surveillance biopsy strategy.
dysplastic Barrett’s esophagus patients after extended follow Gastrointest Endosc 2019;90:395–403.
up; results from Dutch surveillance cohorts. PLoS One 2020;15: 58. Sami SS, Ravindran A, Kahn A, et al. Timeline and location of
e0231419. recurrence following successful ablation in Barrett’s oesoph-
47. Standards of Practice Committee; Wani S, Qumseya B, agus: an international multicentre study. Gut 2019;
Sultan S, et al. Endoscopic eradication therapy for patients with 68:1379–1385.
Barrett’s esophagus-associated dysplasia and intramucosal 59. Komanduri S, Swanson G, Keefer L, et al. Use of a new jumbo
cancer. Gastrointest Endosc 2018;87:907–931.e9. forceps improves tissue acquisition of Barrett’s esophagus sur-
48. Snyder P, Dunbar K, Cipher DJ, et al. Aberrant p53 ommunos- veillance biopsies. Gastrointest Endosc 2009;70:1072–1078.e1.
taining in Barrett’s esophagus predicts neoplastic progression: 60. Kolb JM, Wani S. Endoscopic eradication therapy for Barrett’s
systematic review and meta-analyses. Dig Dis Sci 2019; oesophagus: state of the art. Curr Opin Gastroenterol 2020;
64:1089–1097. 36:351–358.
49. Redston M, Noffsinger A, Kim A, et al. Abnormal TP53 predicts
risk of progression in patients with Barrett’s esophagus
regardless of a diagnosis of dysplasia. Gastroenterology 2022; Correspondence
162:468–481. Address correspondence to: Srinadh Komanduri, MD, MS, Professor of Med-
50. Scholvinck DW, van der Meulen K, Bergman J, et al. Detection icine and Surgery, Associate Chief, Division of Gastroenterology and Hep-
atology, Feinberg School of Medicine, Northwestern University, 676 St Clair,
of lesions in dysplastic Barrett’s esophagus by community and 14th Fl, Chicago, IL 60611. e-mail: koman1973@gmail.com.
expert endoscopists. Endoscopy 2017;49:113–120.
51. Wani S, Muthusamy VR, Shaheen NJ, et al. Development of Acknowledgments
CGIT Barrett’s Esophagus Consensus Conference contributors: Jacques
quality indicators for endoscopic eradication therapies in Bergman, Marcia I. Canto, Amitabh Chak, Douglas Corley, Gary W. Falk,
Barrett’s esophagus: the TREAT-BE (Treatment With Resec- Rebecca Fitzgerald, Rehan Haidry, John M. Haydek, John Inadomi, Prasad G.
Iyer, Vani Konda, Elizabeth Montgomery, Krish Ragunath, Joel Rubenstein,
tion and Endoscopic Ablation Techniques for Barrett’s
Jason B. Samarasena, Felice Schnoll-Sussman, Nicholas J. Shaheen, Michael
Esophagus) Consortium. Am J Gastroenterol 2017; Smith, Rhonda F. Souza, Stuart J. Spechler, Arvind Trindade, Rockford G.
112:1032–1048. Yapp. Please refer to the Supplementary Materials for additional disclosures
and conflicts of interest.
52. Singh S, Garg SK, Singh PP, et al. Acid-suppressive medica-
tions and risk of oesophageal adenocarcinoma in patients with Conflicts of interest
Barrett’s oesophagus: a systematic review and meta-analysis. The authors disclose the following. V. Raman Muthusamy reports consultant
Gut 2014;63:1229–1237. for Medtronic, Boston Scientific, research support from Boston Scientific;
Advisory Board: Endogastric Solutions; honoraria from Torax Medical/Ethicon;
53. Jankowski JAZ, de Caestecker J, Love SB, et al. AspECT Trial and stock from Capsovision. Sachin Wani reports consultant for Exact Sci-
Team. Esomeprazole and aspirin in Barrett’s oesophagus ences advisory board for Cernostics; and research support from Lucid Di-
agnostics and CDx Diagnostics. Prakash Gyawali reports consultant for
(AspECT): a randomised factorial trial. Lancet 2018; Medtronic. Srinadh Komanduri reports consultant for Medtronic, Boston Sci-
392:400–408. entific, Castle Biosciences, and Johnson & Johnson.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.
December 2022 Screening and Surveillance of Barrett’s Esophagus 2706.e1

Supplementary Appendix attached to a thin catheter. The balloon with ridges at the
end of the catheter is inflated and withdrawn for cell
Cell-collection Devices for Barrett’s Esophagus acquisition. The balloon is inverted before withdrawal to
prevent contamination from the squamous mucosa. The
Cytosponge. The Cytosponge is a spherical piece of balloon is placed in a preservative and then evaluated. A
polyurethane foam within a gelatin capsule that is pilot study assessing the EsoCheck device using a 2-
attached to a suture. The capsule is swallowed by the biomarker assay (VIM and CCNA1) demonstrated a
patient; it then dissolves upon reaching the stomach. The sensitivity of 90.3% and specificity of 91.7%.8 A larger
foam then expands and is slowly withdrawn, sampling multicenter study to further validate these findings is
the surface of the gastric cardia and esophagus. The ac- underway. No significant adverse events have been re-
quired cells are placed in a preservative and a paraffin ported though data are limited.
block created. Standard sections are created and stained
with hematoxylin and eosin along with trefoil factor 3, an Supplementary References
immunochemical biomarker for intestinal metaplasia. 1. Kadri SR, Lao-Sirieix P, O’Donovan M, et al. Acceptability and
The Cytosponge has been heavily tested in the United accuracy of a non-endoscopic screening test for Barrett’s
Kingdom in the Barett’s Esophagus (BE) Screening trials oesophagus in primary care: cohort study. BMJ 2010;
(BEST 1-3).1-3 In addition, the Cytosponge has a good 341:c4372.
patient acceptability profile4 and shown in to have a 2. Ross-Innes CS, Debiram-Beecham I, O’Donovan M, et al. ,
BEST2 Study Group. Evaluation of a minimally invasive cell
sensitivity of 80% and specificity of 92%2 for the
sampling device coupled with assessment of trefoil factor 3
detection of BE. In a large, multicenter, pragmatic ran-
expression for diagnosing Barrett’s esophagus: a multi-center
domized controlled trial (BEST 3) in the primary care case-control study. PLoS Med 2015;12:e1001780.
setting, Cytosponge-TFF3 demonstrated a 10-fold in- 3. Fitzgerald RC, di Pietro M, O’Donovan M, et al. BEST3 Trial
crease in detection of BE.3 A rare case of tether detach- team. Cytosponge-trefoil factor 3 versus usual care to identify
ment has been described. The most common adverse Barrett’s oesophagus in a primary care setting: a multicentre,
event has been throat soreness in up to 4%.3 pragmatic, randomised controlled trial. Lancet 2020;
EsophaCap. The EsophaCap or “sponge on a string” 396:333–344.
(SOS) is similar conceptually to the Cytosponge but with 4. Shaheen NJ, Komanduri S, Muthusamy VR, et al. Acceptability
a smaller diameter, at 2.5 cm. The EsophaCap has been and adequacy of a non-endoscopic cell collection device for
studied assessing methylated biomarkers (VAV3, diagnosis of Barrett’s esophagus: lessons learned. Dig Dis Sci
ZNF682) and found to have a 100% sensitivity and 2022;67:177–186.
specificity for intestinal metaplasia (IM).5 This was 5. Iyer PG, Taylor WR, Johnson ML, et al. Highly discriminant
further studied with other methylated biomarkers and methylated DNA markers for the non-endoscopic detection
of Barrett’s esophagus. Am J Gastroenterol 2018;113:1156–
found to have a sensitivity of 94% with specificity of
1166.
62.6%.6 However, both studies were limited in sample
6. Wang Z, Kambhampati S, Cheng Y, et al. Methylation biomarker
size. Ultimately, a 5 methylated DNA marker panel was
panel performance in EsophaCap cytology samples for diag-
validated in a multicenter study using a training (n ¼
nosing Barrett’s esophagus: a prospective validation study. Clin
199) and test cohort (n ¼ 89). Sensitivity of the panel Cancer Res 2019;25:2127–2135.
was 93% for diagnosis of BE with specificity of 90% in 7. Iyer PG, Taylor WR, Slettedahl SW, et al. Validation of a meth-
the training set, whereas the sensitivity was 93% with ylated DNA marker panel for the nonendoscopic detection of
specificity of 93% in the test set.7 The SOS-3 validation Barrett’s esophagus in a multisite case-control study. Gastro-
trial is currently underway. No significant adverse events intest Endosc 2021;94:498–505.
have been reported though data are limited. 8. Moinova HR, LaFramboise T, Lutterbaugh JD, et al. Identifying
EsoCheck. The EsoCheck device is distinct from the DNA methylation biomarkers for non-endoscopic detection of
string-based collection devices utilizing a balloon Barrett’s esophagus. Sci Transl Med 2018;10:eaao5848.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en agosto 09, 2023. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2023. Elsevier Inc. Todos los derechos reservados.

You might also like